WO2017015400A1 - Composés azaaryl substitués par le chlorobenzène - Google Patents

Composés azaaryl substitués par le chlorobenzène Download PDF

Info

Publication number
WO2017015400A1
WO2017015400A1 PCT/US2016/043203 US2016043203W WO2017015400A1 WO 2017015400 A1 WO2017015400 A1 WO 2017015400A1 US 2016043203 W US2016043203 W US 2016043203W WO 2017015400 A1 WO2017015400 A1 WO 2017015400A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
methyl
urea
dimethoxyphenyl
trichloro
Prior art date
Application number
PCT/US2016/043203
Other languages
English (en)
Inventor
Yun Yen
Jing-Ping Liou
Chun-Han Chen
Original Assignee
Taipei Medical University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Taipei Medical University filed Critical Taipei Medical University
Priority to EP16828495.8A priority Critical patent/EP3324970A4/fr
Priority to CA2993022A priority patent/CA2993022A1/fr
Priority to CN201680043128.3A priority patent/CN108348519A/zh
Priority to JP2018502695A priority patent/JP2018527321A/ja
Priority to BR112018001053A priority patent/BR112018001053A2/pt
Priority to AU2016296997A priority patent/AU2016296997B2/en
Priority to US15/746,182 priority patent/US10624897B2/en
Publication of WO2017015400A1 publication Critical patent/WO2017015400A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings

Definitions

  • the invention relates to a series of anti-cancer compounds and the pharmaceutical preparations thereof as well as their methods of use. Particularly, the invention provides chlorobenzene substituted azaaryl compounds having anti-cancer activity.
  • WO 2013144339 relates to 3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-l- ⁇ 6-[4-(4-ethyl-piperazin-l-yl)- phenylamino]-pyrimid-4-yl ⁇ -l-methyl-urea or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition comprising the same for use in the treatment of fibroblast growth factor receptor mediated disorders.
  • One aspect of the invention is to provide a compound having the following Formula (I):
  • Another aspect of the invention is to provide a pharmaceutical composition containing a compound of Formula (I).
  • a further aspect is to provide a method for inhibiting, preventing or treating a cancer, comprising administrating a compound of Formula (I) to a cell or a subject in need thereof.
  • FIGS. 1A and IB show that MPT0L145 inhibits FGFR signaling and exerts anti- growth effects on FGFR-activated cancer cell lines.
  • A Effects of MPT0L145 on the viability of bladder cancer cells.
  • RT-112, RT4 and T24 cells were treated with the indicated concentrations of MPT0L145 for 72 h. Cell viability was assessed the MTT assay. Data are expressed as means ⁇ S.D. (***P ⁇ 0.001 comparing to control group)
  • B. MPT0L145 has less toxicity relative to normal cells. HUVECs were treated with indicated concentrations of MPT0L145 for 72 hours and viability was examined by MTT assay. Data are expressed as means ⁇ S.D. (***P ⁇ 0.001 compares to control group).
  • FIGS. 2A to 2D show the inhibition of FGFR signaling by MPT0L145 in RT-112 cells.
  • B-D Effects of MPT0L145 on FGFR downstream signaling. Cells were treated with the indicated concentrations of MPT0L145 or BGJ-398 for 1 h B. 4 h C. and 8 h D. Protein ly sates were subjected to western blot analysis with the indicated antibodies.
  • FIG. 3A to 3D show effects of MPT0L145 on cell cycle distribution.
  • A. RT-112 cells were treated with MPT0L145 (4 ⁇ ), BGJ-398 (4 ⁇ ) and Paclitaxel (0.1 ⁇ ) for the indicated times, and cell cycle distribution was analyzed via flow cytometry. (CTL: control group, 145: MPT0L145, 398: BGJ-398, Tax: Paclitaxel)
  • CTL control group
  • 145 MPT0L145
  • 398 BGJ-398
  • Tax Paclitaxel
  • C. RT-112 cells were exposed to the indicated concentrations of MPT0L145 and BGJ-398 (8 ⁇ ) for 24 h and subjected to flow cytometry.
  • RT-112 cells were treated with different concentrations of MPT0L145 for 24 h and subjected to western blot. D. RT-112 cells were treated with MPT0L145 and Paclitaxel for 72 h. Apoptosis was assessed via detection of cleaved caspase-3 and PARP.
  • Figure 4A and 4B show the cell viability results (A) and CI values results (B) in A549 cells.
  • Figure 5 A and 5B show the cell viability results (A) and CI values results (B) in Panel cells.
  • the invention provides a series of chlorobenzene substituted azaaryl compounds having activity in inhibiting cancer cell growth and low toxicity to normal cells. Particularly, the compounds of the invention have stronger inhibition effect on bladder cancer and liver cancer.
  • treatment and “treating” embrace both preventative, i.e. prophylactic, or therapeutic, i.e. curative and/or palliative, treatment.
  • treatment and “treating” comprise therapeutic treatment of patients having already developed said condition, in particular in manifest form.
  • Therapeutic treatment may be symptomatic treatment in order to relieve the symptoms of the specific indication or causal treatment in order to reverse or partially reverse the conditions of the indication or to stop or slow down progression of the disease.
  • the compounds, compositions and methods of the present invention may be used for instance as therapeutic treatment over a period of time as well as for chronic therapy.
  • treatment and “treating” comprise prophylactic treatment, i.e. treatment of patients at risk of developing a condition mentioned hereinbefore, thus reducing said risk.
  • terapéuticaally effective amount means an amount of a compound of the present invention that (i) treats or prevents the particular disease or condition, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease or condition, or (iii) prevents or delays the onset of one or more symptoms of the particular disease or condition described herein.
  • substituted means that any one or more hydrogens on the designated atom, radical or moiety are replaced with a selection from the indicated group, provided that the atom's normal valence is not exceeded, and that the substitution results in an acceptably stable compound.
  • pharmaceutically acceptable is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, and commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salts refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof.
  • examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines, pyridine, pyrimidine and quinazoline; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • stereoisomer is a general term for all isomers of individual molecules that differ only in the orientation of their atoms in space. It includes enantiomers and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereoisomers).
  • chiral center refers to a carbon atom to which four different groups are attached.
  • enantiomer and “enantiomeric” refer to a molecule that cannot be superimposed on its mirror image and hence is optically active, wherein the enantiomer rotates the plane of polarized light in one direction and its mirror image compound rotates the plane of polarized light in the opposite direction.
  • racemic refers to a mixture of equal parts of enantiomers which is optically inactive.
  • resolution refers to the separation or concentration or depletion of one of the two enantiomeric forms of a molecule.
  • halo or halogen refers to fluoro, chloro, bromo or iodo.
  • alkyl refers to straight or branched hydrocarbon chains containing the specified number of carbon atoms.
  • C 1 -C6 alkyl is selected from straight-chained and branched non-cyclic hydrocarbons having from 1 to 6 carbon atoms.
  • Representative straight chain C1-C6 alkyl groups include -methyl, -ethyl, -n-propyl, - n-butyl, -n-pentyl, and -n-hexyl.
  • Representative branched C1-C6 alkyls include -isopropyl, - sec-butyl, -isobutyl, -tert-butyl, -isopentyl, -neopentyl, 1 -methylbutyl, 2-methylbutyl, 3- methylbutyl, 1, 1 -dimethylpropyl, 1 ,2-dimethylpropyl, 1-methylpentyl, 2-methylpentyl, 3- methylpentyl, 4-methylpentyl, 1 -ethylbutyl, 2-ethylbutyl, 3-ethylbutyl, 1 , 1-dimethylbutyl, 1,2-dimethylbutyl, 1 ,3-dimethylbutyl, 2,2-dimethylbut l, 2,3-dimethylbutyl, and 3,3- dimethylbutyl.
  • alkenyl refers to straight or branched chain hydrocarbon chains containing the specified number of carbon atoms and one or more double bonds.
  • C2-C6 alkenyl is selected from straight chain and branched non-cyclic hydrocarbons having from 2 to 6 carbon atoms and including at least one carbon-carbon double bond.
  • Representative straight chain and branched C2-C6 alkenyl groups include - vinyl, -allyl, -1 -butenyl, -2-butenyl, -isobutylenyl, -1 -pentenyl, -2-pentenyl, -3-methyl-l - butenyl, -2-methyl-2-butenyl, -2,3-dimethyl-2-butenyl, -1-hexenyl, 2-hexenyl, and 3-hexenyl.
  • a "C2-C6 alkynyl” is selected from straight chain and branched non-cyclic hydrocarbon having from 2 to 6 carbon atoms and including at least one carbon- carbon triple bond.
  • Representative straight chain and branched C2-C6 alkynyl groups include -acetylenyl, -propynyl, -1-butyryl, -2-butyryl, -1-pentynyl, -2-pentynyl, -3-methyl-l -butynyl, -4-pentynyl, -1 -hexynyl, -2-hexynyl, and -5-hexynyl.
  • cycloalkyl refers to a group selected from C3-C12 cycloalkyl, and preferably a C3-8 cycloalkyl.
  • Typical cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and cyclononyl.
  • alkoxy refers to a straight or branched alkoxy group containing the specified number of carbon atoms.
  • Ci- 6 alkoxy means a straight or branched alkoxy group containing at least 1 , and at most 6, carbon atoms.
  • alkoxy as used herein include, but are not limited to, methoxy, ethoxy, propoxy, prop-2- oxy, butoxy, but-2-oxy, 2-methylprop-l-oxy, 2-methylprop-2-oxy, pentoxy and hexyloxy. The point of attachment may be on the oxygen or carbon atom.
  • alkylthio refers to straight- chain or branched alkyl groups (preferably having 1 to 6 carbon atoms, e.g. 1 to 4 carbon atoms (Ci-C6-alkylthio), which are bound to the remainder of the molecule via a sulfur atom at any bond in the alkyl group.
  • Examples of Ci-C4-alkylthio include methylthio, ethylthio, n- propylthio, isopropylthio, n-butylthio, sec-butylthio, isobutylthio and tert-butylthio.
  • Ci-C6-alkylthio include, apart from those mentioned for Ci-C4-alkylthio, 1-, 2- and 3-pentylthio, 1-, 2- and 3-hexylthio and the positional isomers thereof.”
  • alkoxyalkyl refers to the group -alki-0-alk 2 where alki is alkyl or alkenyl, and alk 2 is alkyl or alkenyl.
  • alkylamino refers to the group --NRR' where R is alkyl and R is hydrogen or alkyl.
  • aryl refers to a group selected from C6-14 aryl, especially Ce- ⁇ aryl.
  • Typical Ce-14 aryl groups include phenyl, naphthyl, phenanthryl, anthracyl, indenyl, azulenyl, biphenyl, biphenylenyl and fluorenyl groups.
  • heteroaryl refers to a group having from 5 to 14 ring atoms; 6, 10 or 14 pi electrons shared in a cyclic array; and containing carbon atoms and 1, 2 or 3 oxygen, nitrogen and/or sulfur heteroatoms.
  • heteroaryl groups include indazolyl, furyl, thienyl, pyrrolyl, imidazolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, tetrazolyl, triazinyl, azepinyl, oxazepinyl, morpholinyl, thiazepinyl, diazepinyl, thiazolinyl, benzimidazolyl, benzoxazolyl, imidazopyridinyl, benzoxazinyl, benzothiazinyl, benzothiophenyl oxazolopyridinyl, benzofuranyl, quinolinyl, quina
  • the term "therapeutically effective amount” is meant to refer to an amount of an active agent or combination of agents effective to ameliorate or prevent the symptoms. Determination of a therapeutically effective amount is well within the capabilities of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the invention provides a compounds having the following Formula (I):
  • X is C, N, O or S
  • Ri is cycloalkyl; aryl unsubstituted or substituted by halo, carbonyl, hydroxy, amino, nitro, cyano, alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkoxyalkyl, alkylamino, or heteroaryl having 1 to 3 heteroatoms selected from the group consisting of N, O and S; heteroalkyl unsubstituted or substituted by halo, carbonyl, hydroxy, amino, nitro, cyano, alkoxy, alkylthio, alkoxyalkyl, alkylamino, or heteroaryl having 1 to 3 heteroatoms selected from the group consisting of N, O and S; or NR 7 R 8 wherein R 7 and Rg are each independently selected from the group consisting of H, nitro, amino, cyano, alkyl, alkenyl, alkynyl, aryl or heteroaryl wherein alkyl, alkenyl, alky
  • R-3 and R4 are each independently selected from the group consisting of alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkoxyalkyl or alkylamino;
  • heteroaryl is unsubstituted or substituted by halo, carbonyl, hydroxy, amino, nitro, cyano, alkyl, alkenyl, alkynyl, alkoxy, alkylthio, alkoxyalkyl, alkylamino or aryl;
  • halo is F, CI or Br; alkyl is Ci-ioalkyl, preferably Ci- 6 alkyl or Cl-4alkyl; alkenyl is C 2 -ioalkenyl, preferably C 2 - 6 alkenyl; alkynyl is C 2 -ioalkynyl, preferably C 2 - 6 alkynyl; alkoxy is Ci-ioalkoxy, preferably Ci- 6 alkoxy or Ci_ 4alkoxy; aryl is 5- or 6-membered aryl, preferably phenyl; and heterozryl is 5- or 6-membered heteroaryl and has 1 to 3 heteroatoms selected from the group consisting of N, O and S.
  • X is C; Ri is phenyl, Ci_ l oalkylpiperazinylphenyl, Ci-ioalkyloxyphenyl, halophenyl, cyanophenyl, nitrophenyl, furyl or pyridinyl. More preferably, X is C; Ri is (4-ethylpiperazinyl-l-yl)phenyl, phenyl, 4- methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 4-cyanophenyl, nitrophenyl, 2-furyl, 3- pyridinyl or 4-pyridinyl.
  • Ri is phenyl, (4-ethylpiperazinyl-l-yl)phenyl, phenyl, 4-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl, 4-cyanophenyl, nitrophenyl, 2- furyl, 3-pyridinyl or 4-pyridinyl;
  • R 2 is H;
  • R 3 and R4 are each independently CH 3 ;
  • R5 and R6 are each independently H or Ci-ioalkyl.
  • X is C or N; Ri is NR 7 R 8 wherein R 7 is Ci-ioalkylpiperazinylphenyl, piperidinylCi-ioalkyl, Ci_ l oalkylpiperazinylCl-lOalkyl or Ci-ioalkylpiperazinylcarbonylphenyl and Rg is H; R2 is H, halo or phenyl; R 3 and R4 are each independently Ci-ioalkyl; and R5 and R6 are each independently H or Ci-ioalkyl.
  • the compounds include but are not limited to the following:
  • the compound of the invention is l-(4-((4-(4- ethylpiperazin-l-yl)phenyl)amino)-l,3,5-triazin-2-yl)-l-methyl-3-(2,4,6-trichloro-3,5- dimethoxyphenyl)urea,
  • the invention provides a pharmaceutical composition, comprising a compound of the invention and a pharmaceutically acceptable carrier.
  • the compound of the invention is typically administered in the form of a pharmaceutical composition.
  • Such compositions can be prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
  • the compound of the invention is administered in a pharmaceutically effective amount.
  • the amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • the compound is present in the composition in an amount that is effective to achieve its intended therapeutic purpose. While individual needs may vary, a determination of optimal ranges of effective amounts of each compound is within the skill of the art.
  • the compounds may be administered to a mammal, e.g. a human, orally at a dose of from about 0.1 to about 100 mg per kg body weight of the mammal, or an equivalent amount of a pharmaceutically acceptable salt, prodrug or solvate thereof, per day to treat, prevent or ameliorate the particular disorder.
  • the dose ranges from about 0.1 to about 90 mg, about 0.1 to about 80 mg, about 0.1 to about 70 mg, about 0.1 to about 60 mg, about 0.1 to about 50 mg, about 0.1 to about 40 mg, about 0.1 to about 30 mg, about 0.1 to about 20 mg, about 0.1 to about 10 mg, about 0.1 to about 5 mg, about 0.5 to about 100 mg, about 0.5 to about 90 mg, about 0.5 to about 80 mg, about 0.5 to about 70 mg, about 0.5 to about 60 mg, about 0.5 to about 50 mg, about 0.5 to about 40 mg, about 0.5 to about 30 mg, about 0.5 to about 20 mg, about 0.5 to about 10 mg, about 0.5 to about 5 mg, about 1 to about 100 mg, about 1 to about 90 mg, about 1 to about 80 mg, about 1 to about 70 mg, about 1 to about 60 mg, about 1 to about 50 mg, about 1 to about 40 mg, about 1 to about 30 mg, about 1 to about 20 mg, about 1 to about 10 mg, about 5 to about 100 mg, about 1 to about
  • a useful oral dose of a compound of the present invention administered to a mammal is from about 5 to about 100 mg per kg body weight of the mammal (the preferred dose is as mentioned above), or an equivalent amount of the pharmaceutically acceptable salt, prodrug or solvate thereof.
  • the dose is typically about one-half of the oral dose.
  • a unit oral dose may comprise from about 0.1 to about 100 mg, and preferably about 1 to about 80 mg of a compound.
  • the unit dose can be administered one or more times daily, e.g. as one or more tablets or capsules, each containing from about 0.01 mg to about 50 mg of the compound, or an equivalent amount of a pharmaceutically acceptable salt, prodrug or solvate thereof.
  • compositions of the invention can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intra-articular, intravenous, intramuscular, and intranasal.
  • routes including oral, rectal, transdermal, subcutaneous, intra-articular, intravenous, intramuscular, and intranasal.
  • the compound of this invention is preferably formulated as either an injectable or oral compositions.
  • compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, the compositions are presented in unit dosage forms to facilitate accurate dosing.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient, vehicle or carrier.
  • Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
  • compositions within the scope of the present invention include all compositions where a compound of the present invention is combined with a pharmaceutically acceptable carrier.
  • the compound is present in the composition in an amount that is effective to achieve its intended therapeutic purpose. While individual needs may vary, determination of optimal ranges of effective amounts of each compound is within the skill of the art.
  • compositions of the present invention may be useful in combination with one or more second therapeutic agents, and exemplary pharmaceutical composition can include or exclude any of the one or more second therapeutic agents described herein, particularly therapeutic agents suitable for the treatment and/or prevention of the conditions and diseases presented previously.
  • the second therapeutic agent can be a mitotic inhibitor (such as taxanes (preferably paclitaxel, docetaxel), vinca alkaloids (preferably, vinblastine, vincristine, vindesine and vinorelbine) and vepesid; an anthracycline antibiotic, (such as doxorubicin, daunorubicin, daunorubicin, epirubicin, idarubicin, valrubicin and mitoxantrone); a nucleoside analog (such as gemcitabine); an EGFR inhibitor (such as gefitinib and erlotinib); a folate antimetabolite (such as trimethoprim, pyrimethamine and pemetrexed); cisplatin and carboplatin.
  • a mitotic inhibitor such as taxanes (preferably paclitaxel, docetaxel), vinca alkaloids (preferably, vinblastine, vincristine, vindesine and vinorelbine) and vepe
  • Examples of the second therapeutic agent that can be included or excluded in the exemplary compounds and/or exemplary pharmaceutical compositions can include, but are not limited to, tamoxifen, taxol, vinblastine, etoposide (VP- 16), adriamycin, 5-fluorouracil (5FU), camptothecin, actinomycin-D, mitomycin C, combretastatin(s), more particularly docetaxel (taxotere), cisplatin (CDDP), cyclophosphamide, doxorubicin, methotrexate, paclitaxel and vincristine, and derivatives and prodrugs thereof.
  • Further useful exemplary second therapeutic agents include compounds that interfere with DNA replication, mitosis, chromosomal segregation and/or tubulin activity.
  • Such compounds include adriamycin, also known as doxorubicin, etoposide, verapamil, podophyllotoxin(s), combretastatin(s) and the like.
  • Agents that disrupt the synthesis and fidelity of polynucleotide precursors may also be used.
  • Particularly useful are agents that have undergone extensive testing and are readily available. As such, agents such as 5- fluorouracil (5-FU) are preferentially used in neoplastic tissue, making this agent particularly useful for targeting to neoplastic cells.
  • 5- fluorouracil 5-FU
  • the present invention provides a method for inhibiting, preventing or treating a cancer in a subject, comprising administering to the subject an effective amount of the compound and/or pharmaceutical composition of the invention.
  • Such method includes administering of a compound of the present invention to a subject in an amount sufficient to treat the condition.
  • the cancers include but are not limited to those selected from the group consisting of: neuroblastoma; lung cancer; bile duct cancer; non-small cell lung carcinoma; hepatocellular carcinoma; head and neck squamous cell carcinoma; squamous cell cervical carcinoma; lymphoma; nasopharyngeal carcinoma; gastric cancer; colon cancer; uterine cervical carcinoma; gall bladder cancer; prostate cancer; breast cancer; testicular germ cell tumors; colorectal cancer; glioma; thyroid cancer; basal cell carcinoma; gastrointestinal stromal cancer; hepatoblastoma; endometrial cancer; ovarian cancer; pancreatic cancer; renal cell cancer, Kaposi's sarcoma, chronic leukemia, sarcoma, rectal cancer, throat cancer, melanoma, colon cancer, bladder cancer, mastocytoma, mammary carcinoma, mammary adenocarcinoma, pharyngeal squamous cell carcinoma, testicular cancer
  • the cancer is bladder cancer and hepatocellular carcinoma.
  • the compounds and pharmaceutical compositions of the invention can be used as FGFR inhibitors and thus are effective in treatment or prevention of FGFR-activated cancers. Accordingly, the invention provides a method for inhibiting, preventing or treating a FGFR- activated cancer in a subject, comprising administering to the subject an effective amount of the compound of the invention.
  • suitable pharmaceutical compositions of the invention include powders, granules, pills, tablets, lozenges, chews, gels, and capsules as well as liquids, syrups, suspensions, elixirs, and emulsions. These compositions may also include anti-oxidants, flavorants, preservatives, suspending, thickening and emulsifying agents, colorants, flavoring agents and other pharmaceutically acceptable additives. Formulations for oral administration may be formulated to be immediate release or modified release, where modified release includes delayed, sustained, pulsed, controlled, targeted and programmed release.
  • the compounds and pharmaceutical composition of the present invention are administered directly into the blood stream, into muscle, or into an internal organ via an intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous or other injection or infusion.
  • Parenteral formulations may be prepared in aqueous injection solutions which may contain, in addition to the compound of the invention, buffers, antioxidants, bacteriostats, salts, carbohydrates, and other additives commonly employed in such solutions.
  • Parenteral administrations may be immediate release or modified release (such as an injected or implanted depot).
  • Compounds and pharmaceutical compositions of the present invention may also be administered topically, (intra)dermally, or transdermally to the skin or mucosa.
  • Typical formulations include gels, hydrogels, lotions, solutions, creams, ointments, dressings, foams, skin patches, wafers, implants and microemulsions.
  • Compounds of the present invention may also be administered via inhalation or intanasal administration, such as with a dry powder, an aerosol spray or as drops.
  • Additional routes of administration for exemplary compounds and compositions of the present invention include intravaginal and rectal (by means of a suppository, pessary or enema), ocular and aural.
  • the PdCl 2 dppf (0.04g, 0.06mmol), potassium acetate (0.55g, 5.58mmol) and bispinacolactoboron (0.71g, 2.79mmol) were assed to the mixture of 83 (0.06g, 0.41mmol), H 2 0 (1ml) and p- dioxane (4ml) and the resulting mixture was then stirred and refluxed for overnight. The reaction was quenched by saturated NaHC0 3 (aq.) and the mixture was extracted by ethyl acetate (30ml x 3).
  • Example 42 Biological Assay - MTT Assay [ 0099 ] Cells were cultured in RPMI-1640 (RT-112, KMS-11, SNU-16, HT-29, HCT-116, NCI-H520, Hep3B, PLC/PRF/5, HL-60, MOLT-4) and DMEM (HepG2, MCF-7, MDA-MB- 231) with 10% FBS (v/v) and Penicillin/Streptomycin (lOOU/ml). FU-DDLS-1 cell line was maintained in DMEM:F12 medium with 10% FBS.
  • LiSa-2 cell line was maintained in IMDM /RPMI-1640 in a 4: 1 ratio supplemented with 10% FBS, 2 mmol/L L-glutamine, and 0.1 mg/mL gentamicin.
  • Human umbilical vein endothelial cells (HUVECs) were grown to confluence on 1% collagen, and maintained in 90% Medium 199 with 25 U/ml heparin, 30 ⁇ g/ml endothelial cell growth supplement (ECGS) adjusted to contain 1.5 g/L sodium bicarbonate, 10% FBS and Penicillin/Streptomycin (lOOU/ml) Cultures were maintained at 37°C in a humidified atmosphere of 5% C02/95% air.
  • MTT assay Different cancer cell lines were used in MTT assay to test the anticancer activity of exemplary chlorobenzene substituted azaaryl compound (MPT0L145).
  • a known compound, BJG398 (3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-l- ⁇ 6-[4-(4-ethylpiperazin-l -yl)- phenylamino]-pyrimidin-4-yl ⁇ -l -methyl-urea), was used as reference compound for comparison.
  • BJG398 3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-l- ⁇ 6-[4-(4-ethylpiperazin-l -yl)- phenylamino]-pyrimidin-4-yl ⁇ -l -methyl-urea
  • HCT-116 Colorectal 6.12 ⁇ 0.01 3.98 ⁇ 0.06 HepG2 Liver 1.18 ⁇ 0.16 5.84 ⁇ 0.25 PLC/PRF/5 Liver 1.48 ⁇ 0.30 2.94 ⁇ 0.11
  • MOLT-4 ALL 3.70 ⁇ 0.20 N/A
  • IC50 values of MPT0L145 in RT-112 and HUVEC were 11.1 ⁇ and 0.05 ⁇ , respectively.
  • RT- 112 cells reportedly rely on FGFRs for growth and are therefore chosen to confirm the effects of MPT0L145 on FGFR signaling [22, 23].
  • BGJ-398 a known selective inhibitor of FGFRl to FGFR3, was included as a reference compound.
  • the major downstream pathways of FGFRs are MAPK, PI3K/AKT, and PLC- ⁇ .
  • RT-112 cells which express FGFR3-TACC3, are reportedly unable to activate PLCy due to a deletion of the last exon of FGFR3.
  • MPT0L145 inhibited phosphorylation of ERK at 1 h in a concentration- dependent manner (Figure 2B).
  • the compound displayed better potency than BGJ-398 in inhibiting AKT phosphorylation from 1 to 4 h ( Figure 2B, 2C).
  • the phosphorylation of ERK and AKT were fully repressed by MPT0L145 at 8 h ( Figure 2D).
  • Example 43 MPT0L145 induces cell cycle arrest at the G0/G1 phase in bladder cancer cells
  • mice were inoculated subcutaneously with l x lO 6 RT-112 cells in a total volume of 0.1 mL serum-free medium containing 50% Matrigel (BD Biosciences).
  • MPT0L145 significantly suppressed tumor growth in a dose-dependent manner.
  • the percentages of tumor growth inhibition (% TGI) of cisplatin (5 mg/ kg) and MPT0L145 (5 and 10 mg/kg) are 56.3%, 61.4% and 74.6%, respectively.
  • MPT0L145 exhibited not only comparable antitumor activity to cisplatin, but also better safety, as established from assessment of body weight loss after treatment. Accordingly, we conclude that MPT0L145 possesses significant antitumor activity in vivo.
  • Example 45 In vitro inhibitory effects of MPT0L145 on protein kinases and lipid kinase
  • Kinase assays For most assays, kinase-tagged T7 phage strains were prepared in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage and incubated with shaking at 32°C until lysis. The lysates were centrifuged and filtered to remove cell debris. The remaining kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection.
  • Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase assays.
  • the liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce non-specific binding.
  • Binding reactions were assembled by combining kinases, liganded affinity beads, and test compounds in lx binding buffer (20% SeaBlock, 0.17x PBS, 0.05% Tween 20, 6 mM DTT).
  • BMPR2 >10000
  • PIK3CG >10000
  • IGF1R >10000 ZAP70 >10000
  • FGFR aberrations are common in a wide variety of cancers, such as gene amplifications or activating mutations.
  • the cancers most commonly affected were urothelial (32% FGFR-aberrant); breast (18%); endometrial (13%), squamous lung cancers (13%), and ovarian cancer (9%).
  • urothelial cancers the majority of aberrations were activating mutations in FGFR3, including S249C, R248C, Y373C, G370C, and K650M (Clin Cancer Res. 2016 Jan l ;22(l):259-67.).
  • active FGFR3 mutation In multiple myeloma, active FGFR3 mutation (Y373C, and K650E) are reportedly important in tumor progression (Oncogene. 2001 Jun 14;20(27):3553- 62.).
  • the mutation of FGFR3 (V555M) is also identified as a mechanism of acquire resistance to FGFR inhibitors (Oncogene. 2013 Jun 20;32(25):3059-70.).
  • constitutive activating mutation of the FGFR3b G697C is found in oral squamous cell carcinomas (Int J Cancer. 2005 Oct 20;117(l): 166-8.). Therefore, we examined the inhibitor activity of MPT0L145 on FGFR3 with active mutation.
  • MPT0L145 showed comparable activity in inhibiting FGFR3 (G697C), and with better activity in specifically inhibiting FGFR3 (K650E). The data suggests that MPT0L145 can inhibit not only wild-type FGFR3, but also FGFR3 carrying active mutation (K650E, G697C).
  • MTT assay was used in A549 cells or Panel cell (72h). Cells were seeded in 96- well plates and exposed to DMSO, or indicated compounds for 72 hours. Cell viability was assessed using the 3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay as described previously [46]. Briefly, 100 ⁇ of 0.5 mg/ ml MTT were added to each well and incubated at 37°C for 1 hour. After that, 100 ⁇ of extraction reagents (0.1 M sodium acetate buffer for suspension cells or DMSO for attached cells) were added to each well to lyse cells and absorbance at 550 nm was measured.
  • MTT 3-(4,5-dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide
  • Cell viability was expressed as the percentage of surviving cells in drug-treated versus DMSO-treated control cells (which was considered as 100% viability).
  • concentration that inhibits 50% of cell growth were determined according to the dose-effect curves.
  • the combination index (CI) value was determined from the fraction-affected value of each drug combination according to the Chou- Talalay method by using CompuSyn software (ComboSyn, Inc.), and a combination index value below 1 represents synergism (Pharmacol Rev. 2006 Sep;58(3):621-81.).
  • the cell viability results and CI values results in A549 cells are shown in Figure 4A and 4B, respectively, and are also shown in the table below.

Abstract

L'invention concerne un ensemble de composés azaaryl substitués par le chlorobenzène qui possède une activité inhibitrice de la croissance des cellules cancéreuses et une faible toxicité envers les cellules normales. En particulier, les composés de l'invention ont un fort effet inhibiteur sur le cancer de la vessie et le cancer du foie.
PCT/US2016/043203 2015-07-20 2016-07-20 Composés azaaryl substitués par le chlorobenzène WO2017015400A1 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
EP16828495.8A EP3324970A4 (fr) 2015-07-20 2016-07-20 Composés azaaryl substitués par le chlorobenzène
CA2993022A CA2993022A1 (fr) 2015-07-20 2016-07-20 Composes azaaryl substitues par le chlorobenzene
CN201680043128.3A CN108348519A (zh) 2015-07-20 2016-07-20 经氯苯取代的氮杂芳基化合物
JP2018502695A JP2018527321A (ja) 2015-07-20 2016-07-20 クロロベンゼン置換されたアザアリール化合物
BR112018001053A BR112018001053A2 (pt) 2015-07-20 2016-07-20 compostos, composição farmacêutica e método para inibir, prevenir ou tratar um câncer em um indivíduo
AU2016296997A AU2016296997B2 (en) 2015-07-20 2016-07-20 Chlorobenzene substituted azaaryl compounds
US15/746,182 US10624897B2 (en) 2015-07-20 2016-07-20 Chlorobenzene substituted azaaryl compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562194705P 2015-07-20 2015-07-20
US62/194,705 2015-07-20

Publications (1)

Publication Number Publication Date
WO2017015400A1 true WO2017015400A1 (fr) 2017-01-26

Family

ID=57834598

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/043203 WO2017015400A1 (fr) 2015-07-20 2016-07-20 Composés azaaryl substitués par le chlorobenzène

Country Status (9)

Country Link
US (1) US10624897B2 (fr)
EP (1) EP3324970A4 (fr)
JP (1) JP2018527321A (fr)
CN (1) CN108348519A (fr)
AU (1) AU2016296997B2 (fr)
BR (1) BR112018001053A2 (fr)
CA (1) CA2993022A1 (fr)
TW (1) TWI694067B (fr)
WO (1) WO2017015400A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021063852A1 (fr) * 2019-09-30 2021-04-08 F. Hoffmann-La Roche Ag Pyrimidine substituée pour le traitement et la prophylaxie d'une infection par le virus de l'hépatite b

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110283130B (zh) * 2019-07-05 2023-12-19 温州医科大学 一种1-(2,5-二甲氧基苯基)-3-取代脲类结肠癌抑制剂及其制备和应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090137804A1 (en) * 2004-06-24 2009-05-28 Qiang Ding Compounds and Compositions as Protein Kinase Inhibitors
WO2013144339A1 (fr) * 2012-03-30 2013-10-03 Novartis Ag Inhibiteur de fgfr destiné au traitement de troubles hypophosphatémiques
US8759517B2 (en) * 2005-12-21 2014-06-24 Novartis Ag Pyrirnidinyl aryl urea derivatives being FGF inhibitors

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101483215B1 (ko) * 2010-01-29 2015-01-16 한미약품 주식회사 단백질 키나아제 저해활성을 갖는 비시클릭 헤테로아릴 유도체
PE20160679A1 (es) 2013-10-18 2016-08-04 Eisai Randd Man Co Ltd Inhibidores de pirimidina del fgfr4

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090137804A1 (en) * 2004-06-24 2009-05-28 Qiang Ding Compounds and Compositions as Protein Kinase Inhibitors
US8759517B2 (en) * 2005-12-21 2014-06-24 Novartis Ag Pyrirnidinyl aryl urea derivatives being FGF inhibitors
WO2013144339A1 (fr) * 2012-03-30 2013-10-03 Novartis Ag Inhibiteur de fgfr destiné au traitement de troubles hypophosphatémiques

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3324970A4 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021063852A1 (fr) * 2019-09-30 2021-04-08 F. Hoffmann-La Roche Ag Pyrimidine substituée pour le traitement et la prophylaxie d'une infection par le virus de l'hépatite b

Also Published As

Publication number Publication date
TWI694067B (zh) 2020-05-21
TW201720800A (zh) 2017-06-16
CA2993022A1 (fr) 2017-01-26
EP3324970A1 (fr) 2018-05-30
JP2018527321A (ja) 2018-09-20
AU2016296997B2 (en) 2020-07-09
US20190083499A1 (en) 2019-03-21
CN108348519A (zh) 2018-07-31
AU2016296997A1 (en) 2018-03-08
US10624897B2 (en) 2020-04-21
BR112018001053A2 (pt) 2018-09-11
EP3324970A4 (fr) 2019-03-06

Similar Documents

Publication Publication Date Title
Singh et al. Targeting non-small cell lung cancer with small-molecule EGFR tyrosine kinase inhibitors
CN109152784B (zh) 经取代的menin-mll抑制剂及使用方法
KR101656382B1 (ko) 피리미딘-2,4-디아민 유도체 및 이를 유효성분으로 함유하는 항암용 약학적 조성물
KR101668931B1 (ko) 항종양제의 효과 증강제
KR20080004495A (ko) 암을 치료하기 위한 조합물, 방법 및 조성물
Liang et al. An update of new small-molecule anticancer drugs approved from 2015 to 2020
US10245251B2 (en) Synergistic pharmaceutical combination for the treatment of squamous cell carcinoma of head and neck
CA3180686A1 (fr) Nouvelles petites molecules pour la degradation ciblee de kras ne pouvant etre cible dans une therapie anticancereuse
JP2019514869A (ja) ErbB阻害剤及びその使用
CN117715905A (zh) 检查点激酶1(chk1)抑制剂及其用途
JP2023554471A (ja) Kras g12c阻害剤を含む医薬組み合わせ及び癌の治療のためのkras g12c阻害剤の使用
CN110831596A (zh) 作为蛋白激酶抑制剂的氨基噻唑化合物
AU2016296997B2 (en) Chlorobenzene substituted azaaryl compounds
JP7372253B2 (ja) 癌の治療における使用のためのチューブリン重合阻害剤とポリ(adp-リボース)ポリメラーゼ(parp)阻害剤との新規組合せ
EP3731845A1 (fr) Composés hétérocycliques en tant que famille (tam) tyro3, axl et mertk d'inhibiteurs de récepteur tyrosine kinase
JP2018083801A (ja) 5’−ヒドロキシ−5−ニトロ−インジルビン−3’−オキシムを有効成分として含有する乳がん治療剤
WO2014002922A1 (fr) Méthode de traitement du cancer par l'utilisation combinée d'un agent anti-cancéreux
JP2014034531A (ja) Hsp90阻害剤とゲムシタビンの組み合わせ
RU2425677C2 (ru) Соединения, предназначенные для использования в фармацевтике
TW201825092A (zh) 使用包含吲哚嗪并[6, 7-b]吲哚衍生物的醫藥組合物或組合治療小細胞肺癌的方法
TW201605449A (zh) 馬賽替尼治療結腸直腸癌的用途
JP2014091711A (ja) Hsp90阻害剤と抗腫瘍性白金錯体との組み合わせ
JP2014034533A (ja) Hsp90阻害剤とegfrチロシンキナーゼ阻害剤の組み合わせ
JP2015163591A (ja) 抗癌剤の併用による癌治療方法

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16828495

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2993022

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2018502695

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2016296997

Country of ref document: AU

Date of ref document: 20160720

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112018001053

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112018001053

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20180118